Next Article in Journal
Early Postoperative Death in Patients Undergoing Emergency High-Risk Surgery: Towards a Better Understanding of Patients for Whom Surgery May not Be Beneficial
Next Article in Special Issue
New Treatment Options for Hyperkalemia in Patients with Chronic Kidney Disease
Previous Article in Journal
Compliance of Atrial Fibrillation Treatment with the Atrial Fibrillation Better Care (ABC) Pathway Improves the Clinical Outcomes in the Middle East Population: A Report from the Gulf Survey of Atrial Fibrillation Events (SAFE) Registry
Previous Article in Special Issue
GLP-1 Receptor Agonists and Diabetic Kidney Disease: A Call of Attention to Nephrologists
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Calciprotein Particles and Serum Calcification Propensity: Hallmarks of Vascular Calcifications in Patients with Chronic Kidney Disease

by
Ciprian N. Silaghi
1,*,
Tamás Ilyés
1,
Adriana J. Van Ballegooijen
2 and
Alexandra M. Crăciun
1
1
Department of Molecular Sciences, University of Medicine and Pharmacy “Iuliu Hațieganu”, 400012 Cluj-Napoca, Romania
2
Department of Nephrology & Epidemiology and Biostatistics, Amsterdam UMC, location VUmc, 1117 HV Amsterdam, Netherlands
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2020, 9(5), 1287; https://doi.org/10.3390/jcm9051287
Submission received: 1 April 2020 / Revised: 27 April 2020 / Accepted: 29 April 2020 / Published: 29 April 2020
(This article belongs to the Special Issue A Changing Perspective for Treatment of Chronic Kidney Disease)

Abstract

:
Cardiovascular complications are one of the leading causes of mortality worldwide and are strongly associated with atherosclerosis and vascular calcification (VC). Patients with chronic kidney disease (CKD) have a higher prevalence of VC as renal function declines, which will result in increased mortality. Serum calciprotein particles (CPPs) are colloidal nanoparticles that have a prominent role in the initiation and progression of VC. The T50 test is a novel test that measures the conversion of primary to secondary calciprotein particles indicating the tendency of serum to calcify. Therefore, we accomplished a comprehensive review as the first integrated approach to clarify fundamental aspects that influence serum CPP levels and T50, and to explore the effects of CPP and calcification propensity on various chronic disease outcomes. In addition, new topics were raised regarding possible clinical uses of T50 in the assessment of VC, particularly in patients with CKD, including possible opportunities in VC management. The relationships between serum calcification propensity and cardiovascular and all-cause mortality were also addressed. The review is the outcome of a comprehensive search on available literature and could open new directions to control VC.

1. Introduction

Serum calciprotein particles (CPPs) are colloidal nanoparticles comprising a combination of proteins (mainly fetuin-A, but also albumin and Gla-rich protein (GRP)) and calcium (Ca2+) containing compounds, primarily calcium phosphate [1,2,3]. They are first formed by the binding of Ca2+ precursors to the acidic residues of fetuin-A, a glycoprotein secreted by the liver [1,4]. These calcium–protein complexes, also known as calciprotein monomers, pass through further aggregation and maturation, resulting in primary calciprotein particles (CPP I) and later on, secondary calciprotein particles (CPP II) [5,6,7]. CPP I are small spherical colloidal nanoparticles that contain amorphous calcium phosphate, while CPP II contain crystalline calcium phosphate at their core, are larger than CPP I, and have a needle-shaped structure. This transition from CPP I to CPP II is called “ripening” and is hypothesized to be attributed to a reorganization of the colloidal nanoparticles into a more stable form [5]. The ripening process is influenced by a number of factors such as the concentration of fetuin-A, Ca2+, magnesium (Mg2+), phosphate (Pi), as well as the temperature and pH of the surrounding microenvironment [1,6,8].
The transition from CPP I to CPP II, which takes place naturally in serum, can also be induced in vitro, and the time needed for the transition to take place can be measured. Half of the time needed for the spontaneous transition from CPP I to CPP II, designated as T50, has been established as a strong predictor of the calcifying properties of serum [9]. A higher T50 is beneficial since serum with a higher T50 is less prone to calcify tissues compared to serum that has a lower T50.
Vascular calcification (VC) results in the thickening and increased rigidity of muscular arterial walls [10]. This is the consequence of two main types of calcification: intimal and medial calcification. Intimal calcification is associated with atherosclerosis, Ca2+ being deposited along with lipoproteins as well as phospholipids [11,12]. Medial calcification, which is more prevalent in chronic kidney disease (CKD), is the result of an osteogenic process similar to intramembranous ossification, which is independent of atherosclerosis and causes a decrease in compliance of the vessel wall [13,14,15]. Medial calcification occurs earlier in CKD patients compared to the general population [16].
With respect to CPPs in general and T50 in particular, there have been no reviews published until now that summarize findings related to both CPPs and T50. Therefore, the purpose of this review was to offer a synopsis of all studies published on CPPs and T50, respectively. We also aim to analyse and discuss their roles and clinical significance in patients prone to developing VC, as well as to establish possible new directions in the management of VC.

2. Methodology

2.1. Search Strategy

All databases that could be accessed through the PubMed search engine were selected for this review. Human, animal, and in vitro studies were all taken into account. Due to the specific nature of the selected domain and the fact that the majority of research papers were published relatively recently, the period of publication was not limited. A set of search terms was selected as follows: “Calciprotein particles”, “T50 AND calcification”, “Serum calcification propensity”. The search was performed in PubMed on the 4th of January 2020 for both search strings, yielding a total of 162 studies (78, 30, and 54 results, respectively). The results of the searches were organized into lists that were cross-checked between search terms, with duplicates being eliminated. After the initial screening of titles and abstracts, full-text articles were obtained for all eligible studies.

2.2. Selection, Screening, and Inclusion

The authors jointly selected the inclusion and exclusion criteria. Only articles with abstracts were selected for screening, written in English including human, animal, and in vitro studies.
Studies that did not address CPPs and/or T50 in a medically relevant manner, such as physical or chemical characterization of CPPs, and studies that lacked a clear definition of methods and materials were not included. Reviews and case reports were excluded as well.
The identification, selection, screening, and inclusion process is summarized in Figure 1. After cross-checking and eliminating duplicates, the results of the search string “Serum calcification propensity” yielded three studies that were subsequently included in the same category as T50. In total, 18 studies were included for CPPs [3,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33] and 30, including the aforementioned 3 studies, for T50 [34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].

3. Molecular Background

3.1. Fetuin-A and Calciprotein Particles

While CPPs contain a number of proteins that can bind Ca2+, e.g., Gla-rich protein (GRP) [3], as well as other serum proteins and lipoproteins such as albumin and apolipoprotein A1 [2], the main protein within the CPP structure is fetuin-A, also known as alpha-2-HS-glycoprotein. It is a 55–60 kDa glycoprotein, synthetized and secreted by the liver, which undergoes post-translational modifications, including phosphorylation [4,64]. While phosphorylation is crucial for its various interactions, e.g., with the insulin receptor, it is not required for mineral binding due to the number of acidic residues [1,4,65,66]. Each molecule of fetuin-A can bind up to 6 Ca2+ ions [67]. Calcium and Pi bound by fetuin-A form protein–mineral complexes called calciprotein monomers, the aggregation of which results in the formation of plasma-soluble amorphous colloidal particles, referred to as CPP I. The CPP I, which is spherical in nature and has a diameter of around 75 nm, circulates in plasma and eventually undergoes rearrangement into CPP II, which is more dense, with a larger diameter (120 nm), insoluble in serum, and has a needle-shaped crystalline structure [1]. This transition from the primary, more instable form, to the secondary, more stable form, is dubbed “ripening” [5]. The process is illustrated in Figure 2.
CPP I and CPP II are cleared by macrophages, especially Kupffer cells in the liver, thereby preventing tissular deposition of Ca2+ and Pi [68]. Studies have shown that CPP II induces vascular smooth muscle cell (VSMC) calcification in vitro, as well as the secretion of tumour necrosis factor α (TNF-α) in macrophages, while CPP I does not. CPP II was found to increase bone morphogenetic protein-2 as well as nuclear factor kappa-B expression in VSMCs. The calcification of VSMCs was also shown to be the result of the cellular uptake of CPP II, with CPP II being detected intracellularly in calcified VSMCs [29]. Both CPP I and CPP II were found to induce VSMC intimal hyperplasia, which was more pronounced in the case of CPP II [18]. Moreover, CPPs were found to induce secretion of interleukin 1β (IL-1β) in macrophages, however, to a lesser degree than hydroxyapatite crystals [31]. While both forms of CPP have pro-inflammatory effects, it is still less prominent than crystalline hydroxyapatite. The more pronounced pro-inflammatory effect of CPP II compared to that of CPP I might be attributed to its content of hydroxyapatite in crystalline form.
The CPPs are detected and quantified in serum indirectly, by assessing the fetuin-A levels via enzyme-linked immunosorbent assay (ELISA), before and after a high-speed centrifugation that precipitates all CPPs as CPP II. The difference between fetuin-A concentrations before and after centrifugation is interpreted as the amount of CPPs in the serum sample [33,69]. Because this method induces the ripening process before measuring CPP content, it only brings information regarding the total concentration of CPPs, without differentiating between CPP I and CPP II. To measure CPP I and CPP II concentrations independently, a flow-cytometry method can be used [70].

3.2. Calcifying Properties of Serum

A method for measuring the calcification inhibition capacity of serum was elaborated by Ismail et al. [71] based on electrochemical impedance. A prototype probe was successfully used to measure the impedance of a test solution consisting of bovine albumin, Ca2+, and Pi. Upon the addition of a calcification inhibitor, in that case fetuin-A, the electrical impedance of the solution would increase proportionately to the Ca2+ content, due to the inhibitor consuming Ca2+ ions by forming CPP I. Thus, the calcification inhibition capacity of the serum could be determined by measuring the variation of impedance of a solution containing Ca2+ and Pi in a known concentration, after the addition of serum.
Pasch et al. [9] were the first to develop a plate-based nephelometric assay to measure the time needed for the transition from CPP I to CPP II in serum treated with Ca2+ and Pi solutions, and proposed the use of one half of the transition time to maximum turbidity, also known as T50, as a parameter to describe the calcifying properties of serum. The influence of factors such as pH and concentrations of various serum constituents upon T50 was also analysed, and is summarized in Figure 3.

4. Results

4.1. Calciprotein Particles

Human studies on serum CPP levels are summarized in Table 1, animal and in vitro studies on CPP are summarized in Table 2. The majority of studies used detection methods that did not differentiate between the two types of CPP. To avoid confusion, we used the term total CPP (tCPP) when referring to studies that did not specify the type of CPP analysed.
Dialysate from haemodialysis (HD) patients was found to contain CPP, and higher dialysate Ca2+ content was found to be associated with higher CPP concentration [24,30]. This suggests that CPP can be cleared from the plasma of patients with chronic kidney disease (CKD) through HD. In addition, CPP were found to induce VSMC calcification and intimal hyperplasia, with higher serum levels of CPP being associated with increased aortic stiffness [18,26,33]. CPP also induced the secretion of TNF-α and IL-1β in macrophages, with a more pronounced effect being attributed to CPP II. This pro-inflammatory response, however, was still inferior to that induced by pure hydroxyapatite crystals [29,31].

4.2. Calcification Propensity

Observational studies of T50 and outcomes are summarized in Table 3, and human intervention studies are summarized in Table 4. The majority of studies included in this section concern T50 in CKD and/or kidney transplant patients.
Oral Mg2+ supplementation, as well as increased Mg2+ concentration in dialysis solution was found to increase T50 in CKD patients [39,45,51]. The T50 was also found to be associated with serum Mg2+ levels in CKD patients, but not with eGFR [50]. Serum Mg2+ levels were directly associated with T50, which suggests that both oral Mg2+ supplementation, as well as increasing the Mg2+ content of dialysis solution could be a viable method to counterbalance VC to some extent in CKD patients. The use of citrate-buffered dialysis solution was found to significantly increase T50 as opposed to standard acetate-buffered dialysis solution in HD patients [34,46]. While platelet derived growth factor B hypomorphic animal brains showed signs of calcification, T50 did not differ compared to controls [61].
Lower T50 levels were also found to be associated with lower tissue oxygenation, as well as an increase in all-cause and cardiovascular mortality, especially in CKD and kidney transplant patients [36,42,48,49,52,56,57,60].

5. Discussion

This comprehensive review showed that multiple lines of evidence (cell, animal, and human) indicate that T50 is shorter in CKD and dialysis populations. A large amount of studies indicate that a lower T50 is related to VC, cardiovascular events, and mortality. These findings are robust across various populations and open up new directions to modify VC especially in patients with CKD. One of these factors that can influence the tendency to calcify is Mg2+. Oral Mg2+ supplementation as well as increased dialysis solution Mg2+ concentration had beneficial effects on T50 [39,45,52], and a lower T50 was associated with cardiovascular and all-cause mortality in various populations [36,42,49,52,56,57,60]. It is worth noting the correlation between higher serum CPP content, especially CPP II, and VSMC inflammation as well as calcification [18,26,29,31,33]. Taking the included studies into consideration, we address two topics for further research in this relatively recent domain.

5.1. The Effect of Dialysis Solution Composition upon Serum Calcification Propensity in CKD Patients

The transition from CPP I to CPP II is delayed by the presence of Mg2+, this effect being dependent upon the concentration of Mg2+. The presence of Mg2+, however, does not inhibit VSMC calcification in the presence of CPP II, suggesting that the anti-calcific effects of Mg2+ are more related to preventing the transition from CPP I to CPP II [19]. This would also explain the effect of Mg2+ upon increasing T50. However, the exact mechanism by which Mg2+ inhibits the maturation of CPP I is not completely understood. One possible mechanism might lie in the ability of Mg2+ to inhibit Ca2+ and Pi crystallization [72], which is a necessary step in CPP maturation.
Studies suggest that there is a significant amount of CPPs in the dialysate of CKD patients on peritoneal dialysis. That CPP content was also directly proportional to the dialysate’s Ca2+ content [24]. While HD was found to increase T50, thus reducing the calcification propensity of the patient’s plasma [37,54], serum CPP I and CPP II levels seem to be unaffected by standard HD [17].
First of all, this would suggest that the increase in T50 after initiation of HD is not attributed to the clearance of CPPs per se, but to the reduction of factors that precipitate the ripening process, most probably the reduction of Ca2+ and Pi. Second of all, CPPs, while not being cleared from the serum under standard HD conditions, are cleared by peritoneal dialysis to some degree. However, if the Mg2+ concentration of HD dialysis solution is increased, CPPs appear to pass the dialysis membrane and are cleared from the patient’s serum [17]. This would, in part, explain the significant increase of T50 in patients treated with a dialysis solution containing a larger Mg2+ concentration compared to standard solution [45].
In addition to the beneficial effect of increased Mg2+ content in dialysis solution upon the serum calcification propensity in CKD patients, the use of an acetate-free, citrate-acidified dialysis solution was also found to increase T50 thus reducing the calcification propensity [34,46].
Patients with CKD who received oral Mg2+ supplementation showed a significant increase in T50 [39,51]. In post-menopausal women, the introduction of oral Ca2+ supplementation showed a decrease in T50, however, this decrease did not differ significantly from the control group [55]. These observations correspond with the findings of Pasch et al. [9], who determined that higher serum Mg2+ levels will increase T50. A summary of the aforementioned factors upon T50 is presented in Figure 4.
Furthermore, it is well known that patients with CKD have a significantly higher risk for VC and associated cardiovascular mortality [73]. Developing a standardized treatment plan for end-stage CKD patients on HD or peritoneal dialysis that would take into account the above outlined criteria, namely the increased Mg2+ content of dialysis solution and the use of citrate instead of acetate, should be validated and subsequently introduced into a therapeutic protocol. Patients with HD, as well as those with CKD who do not require HD, could also benefit from a reduction in oral Ca2+ and an increase in oral Mg2+ supplementation, respectively. Such an approach to the management of VC and the possible ensuing reduction of cardiovascular mortality rates in CKD patients could lead to an increased quality of life, especially for patients undergoing HD or peritoneal dialysis, delaying the onset or decreasing the severity of cardiovascular complications associated with CKD.

5.2. The T50 Test Could Be Used as a Factor in the Staging and/or Prognosis of CKD

There are plentiful studies, conducted on large sample sizes, that came to the conclusion that lower T50 corresponding to higher calcification propensity is strongly associated with increased cardiovascular and all-cause mortality rates [36,42,49,52,56,57,60]. Lower T50 was also associated with coronary artery calcification progression as well as greater risk for cardiovascular disease outcomes, such as myocardial infarction and peripheral vascular events [38,42,49].
The investigation of a possible association between T50 and eGFR could lead to the development of a reference interval for T50 in CKD patients, which is dependent on CKD stage. Such a reference interval, which has not yet been established, could be used as an additional prognostic parameter for CKD patients, especially those undergoing HD or peritoneal dialysis treatment. There was conflicting evidence that links serum CPP levels and T50 to eGFR. Yamada et al. [28] found that CPP levels were inversely associated with eGFR in diabetic patients. However, that study was conducted on diabetic patients, not CKD patients, and the patient group was relatively small as well. On the other hand, Bielesz et al. [50], found that T50 was not associated with eGFR in CKD stage I–V patients, instead being associated with numerous parameters, including Pi and Mg2+ levels. A similar result was obtained by de Seigneux et al. [59], who discovered that T50 was independent of eGFR in kidney transplant donors, which could be attributed to the compensation effect of an otherwise healthy remaining kidney. Those studies clearly pointed that while serum CPP levels are correlated with eGFR, T50 was not.
The CPP levels and T50 do not seem to be directly correlated with one another, although T50 is greatly influenced by serum Ca2+ levels and, in addition, CPP levels are directly proportional to circulating Ca2+ levels. Considering the previously discussed ideas, it could be hypothesized that CPP levels are correlated with T50, justifying further studies in larger populations to investigate the association between T50 and eGFR. However, until the completion of this review, no studies have identified this relationship.
Even in the absence of a link between T50 and eGFR, but in the context of association between higher serum calcification propensity and increased cardiovascular and all-cause mortality rates especially in CKD patients, the use of T50 as risk factor that can be monitored should be considered. The ensuing introduction of measures to decrease calcification propensity could significantly reduce VC and related mortality in CKD patients.
An interesting opportunity would be to expand the area of research towards the involvement of CPPs in the calcification paradox, in which the presence of vascular calcification overlaps at the same time with bone demineralization assessed by a decrease in bone mineral density (BMD) [74]. It is difficult to decode how CPP and the interplay between vasculature–bone–kidney underlie the deleterious effect of calcification. On one hand, fetuin-A accumulates in calcified atherosclerotic plaques [75], but also in bone where it inhibits mineralization and halts bone matrix protein expression [76]. On the other hand, serum levels of fetuin-A were found to be decreased in patients with end-stage renal disease [77]. Contrariwise, serum CPP increases in patients with CKD III–IV, with it being the highest in HD patients [32] but with less fetuin-A content as CKD stage worsens [3]. Probably the turn-over of CPP is accelerated in CKD patients, but fetuin-A is consumed exerting its systemic anti-calcification effect necessary to counteract VC as CKD stage aggravates.
In addition to the well-known presence of VC in patients with CKD, an important decrease of BMD was also reported [78]. In maintenance HD patients, serum fetuin-A was inversely associated with coronary artery calcification and positively with BMD [79]. In respect to VC, serum CPP appears to behave divergently regarding fetuin-A dynamics in CKD patients: higher CPP levels are associated with increased aortic stiffness [33] and larger CPP II diameters were found in patients with VC [23]. As might be expected, T50 was inversely associated with coronary artery calcification (CAC) severity in CKD patients [38], thereby, the T50 test seems to mimic serum fetuin-A variations in respect to VC, as they were found to be associated [50]. Regarding the loss of skeletal mineral, T50 was not associated with BMD [38] and in the case of CPPs we did not find conclusive studies. To make the puzzle even more complicated, we could introduce the relationship between CPPs or T50 and eGFR, as discussed above. Thereby, CPPs were found to be inversely correlated with eGFR [28], instead of T50, which was independent of eGFR [50,59].
However, an attempt to explain the paradox of calcification on the vasculature–bone–kidney axis only in terms of fetuin-A content of CPPs is an exercise of simplification. Given this standpoint, more targeted studies are needed to demonstrate that CPPs are more likely to hold the key on how physiological ossification has correspondence with pathological calcification.
Nevertheless, we need to take into account the current limitations of the T50 test. Several weaknesses were identified by Pasch et al. [9]: the test overrides the contribution of VSMCs and calcifying myeloid cells in promoting VC in vivo, and the serum pH had no influence on the test due to the presence of a strong buffer. Another issue is attaining standardized conditions to perform the test. Consequently, even if a reference interval would be preferable to be established, each laboratory is likely to set up its own different reference interval, hence it is hard to envisage an accepted consensus interval. The test is robust but needs further development in terms of time per test which is too long to be clinically implemented for the moment: to perform a 96-well format takes 10 h [9].
In addition, simply minimizing the T50 as a marker only for VC may be incomplete. The T50 could be considered as a momentary status of the sum of pro- and anti-calcification factors in the serum of a patient, but this may have implications on other pathophysiological processes, thus opening a wide field of research. Accordingly, the term mineral stress has been coined by Pasch et al. [80] and refers to the interaction between inflammation, oxidative stress, and calcification promoted by CPP II.

6. Conclusions

The relatively recent discovery of CPPs opens up new possibilities for the prevention of VC and the attempt to quantify the serum calcification propensity via T50. Even though the factors that influence serum CPP levels, including their ripening process, as well the effect of various factors upon T50 and its variation in different diseases is incompletely understood, there is mounting evidence suggesting that T50 could be a viable marker in the assessment of VC. Moreover, T50 could be valuable in managing VC in CKD patients, especially those undergoing HD, who have a significantly increased risk for developing cardiovascular complications. In these situations, the early introduction of a treatment strategy that increases T50 could mitigate the obvious complications related to VC. Such an approach is still at an early phase, warranting future studies on the use of T50 as a standard tool in the assessment of VC, thus allowing early measures to prevent cardiovascular complications in patients at risk.

Abbreviations:

BMDBone Mineral Density
Ca2+Calcium
CACCoronary Artery Calcification
CKDChronic Kidney Disease
CPP IPrimary Calciprotein Particles
CPP IISecondary Calciprotein Particles
CPPCalciprotein Particles
CVDCardiovascular Disease
CVECardiovascular Event
eGFREstimated Glomerular Filtration Rate
ELISAEnzyme-Linked Immunosorbent Assay
GRPGla-Rich Protein
HDHaemodialysis
H2SHydrogen Sulphide
IL-1βInterleukin 1β
Mg2+Magnesium
N/ANot Applicable
PiPhosphate
SLESystemic Lupus Erythematosus
tCPPTotal Calciprotein Particles
TNF-αTumour Necrosis Factor α
TODTime of Death
VCVascular Calcification
VSMCVascular Smooth Muscle Cell

Author Contributions

All authors have contributed equally to this manuscript. Conceptualization, C.N.S. and T.I.; methodology, T.I.; Writing—original draft, T.I. and C.N.S.; Writing—review and editing, A.J.v.B. and A.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant of the Ministry of Research and Innovation, CNCS—UEFISCDI, project number PN-III-P4-ID-PCCF-2016-0016, within PNCDI III.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Heiss, A.; Duchesne, A.; Denecke, B.; Grötzinger, J.; Yamamoto, K.; Renné, T.; Jahnen-Dechent, W. Structural Basis of Calcification Inhibition by α2-HS Glycoprotein/Fetuin-A. J. Boil. Chem. 2003, 278, 13333–13341. [Google Scholar] [CrossRef] [Green Version]
  2. Köppert, S.; Büscher, A.; Babler, A.; Ghallab, A.; Buhl, E.M.; Latz, E.; Hengstler, J.G.; Smith, E.R.; Jahnen-Dechent, W. Cellular Clearance and Biological Activity of Calciprotein Particles Depend on Their Maturation State and Crystallinity. Front. Immunol. 2018, 9, 9. [Google Scholar] [CrossRef]
  3. Viegas, C.S.; Santos, L.; Macedo, A.; Matos, A.P.; Silva, A.P.; Neves, P.L.; Staes, A.; Gevaert, K.; Morais, R.; Vermeer, C.; et al. Chronic Kidney Disease Circulating Calciprotein Particles and Extracellular Vesicles Promote Vascular Calcification. Arter. Thromb. Vasc. Boil. 2018, 38, 575–587. [Google Scholar] [CrossRef] [Green Version]
  4. Schinke, T.; Amendt, C.; Trindl, A.; Pöschke, O.; Müller-Esterl, W.; Jahnen-Dechent, W. The Serum Protein α2-HS Glycoprotein/Fetuin Inhibits Apatite Formationin Vitroand in Mineralizing Calvaria Cells. J. Boil. Chem. 1996, 271, 20789–20796. [Google Scholar] [CrossRef] [Green Version]
  5. Holt, S.G.; Smith, E.R. Fetuin-A-containing calciprotein particles in mineral trafficking and vascular disease. Nephrol. Dial. Transplant. 2016, 31, 1583–1587. [Google Scholar] [CrossRef] [Green Version]
  6. Heiss, A.; Eckert, T.; Aretz, A.; Richtering, W.; Van Dorp, W.; Schäfer, C.; Jahnen-Dechent, W. Hierarchical Role of Fetuin-A and Acidic Serum Proteins in the Formation and Stabilization of Calcium Phosphate Particles. J. Boil. Chem. 2008, 283, 14815–14825. [Google Scholar] [CrossRef] [Green Version]
  7. Cai, M.M.X.; Smith, E.R.; Holt, S.G. The role of fetuin-A in mineral trafficking and deposition. BoneKey Rep. 2015, 4, 672. [Google Scholar] [CrossRef] [Green Version]
  8. Rochette, C.N.; Rosenfeldt, S.; Heiss, A.; Narayanan, T.; Ballauff, M.; Jahnen-Dechent, W. A Shielding Topology Stabilizes the Early Stage Protein-Mineral Complexes of Fetuin-A and Calcium Phosphate: A Time-Resolved Small-Angle X-ray Study. ChemBioChem 2009, 10, 735–740. [Google Scholar] [CrossRef]
  9. Pasch, A.; Farese, S.; Gräber, S.; Wald, J.; Richtering, W.; Floege, J.; Jahnen-Dechent, W. Nanoparticle-Based Test Measures Overall Propensity for Calcification in Serum. J. Am. Soc. Nephrol. 2012, 23, 1744–1752. [Google Scholar] [CrossRef]
  10. Hunt, J.L.; Fairman, R.; Mitchell, M.E.; Carpenter, J.P.; Golden, M.; Khalapyan, T.; Wolfe, M.; Neschis, D.; Milner, R.; Scoll, B.; et al. Bone Formation in Carotid Plaques. Stroke 2002, 33, 1214–1219. [Google Scholar] [CrossRef] [Green Version]
  11. Van Oostrom, O.; Fledderus, J.O.; De Kleijn, D.; Pasterkamp, G.; Verhaar, M. Smooth Muscle Progenitor Cells: Friend or Foe in Vascular Disease? Curr. Stem Cell Res. Ther. 2009, 4, 131–140. [Google Scholar] [CrossRef] [Green Version]
  12. Persy, V.; D’Haese, P. Vascular calcification and bone disease: The calcification paradox. Trends Mol. Med. 2009, 15, 405–416. [Google Scholar] [CrossRef]
  13. Ho, C.Y.; Shanahan, C.M. Medial Arterial Calcification. Arter. Thromb. Vasc. Boil. 2016, 36, 1475–1482. [Google Scholar] [CrossRef] [Green Version]
  14. Lanzer, P.; Boehm, M.; Sorribas, V.; Thiriet, M.; Janzen, J.; Zeller, T.; Hilaire, C.S.; Shanahan, C.M. Medial vascular calcification revisited: Review and perspectives. Eur. Hear. J. 2014, 35, 1515–1525. [Google Scholar] [CrossRef]
  15. Magne, D.; Julien, M.; Vinatier, C.; Weiss, P.; Guicheux, J.; Merhi-Soussi, F. Cartilage formation in growth plate and arteries: From physiology to pathology. BioEssays 2005, 27, 708–716. [Google Scholar] [CrossRef]
  16. Goodman, W.G.; Goldin, J.; Kuizon, B.D.; Yoon, C.; Gales, B.; Sider, D.; Wang, Y.; Chung, J.; Emerick, A.; Greaser, L.; et al. Coronary-Artery Calcification in Young Adults with End-Stage Renal Disease Who Are Undergoing Dialysis. New Engl. J. Med. 2000, 342, 1478–1483. [Google Scholar] [CrossRef]
  17. Bressendorff, I.; Hansen, D.; Pasch, A.; Holt, S.G.; Schou, M.; Brandi, L.; Smith, E.R. The effect of increasing dialysate magnesium on calciprotein particles, inflammation and bone markers: Post hoc analysis from a randomized controlled clinical trial. Nephrol. Dial. Transplant. 2019. [Google Scholar] [CrossRef]
  18. Shishkova, D.; Velikanova, E.; Sinitsky, M.; Tsepokina, A.; Gruzdeva, O.V.; Bogdanov, L.; Kutikhin, A. Calcium Phosphate Bions Cause Intimal Hyperplasia in Intact Aortas of Normolipidemic Rats through Endothelial Injury. Int. J. Mol. Sci. 2019, 20, 5728. [Google Scholar] [CrossRef] [Green Version]
  19. Ter Braake, A.D.; Eelderink, C.; Zeper, L.W.; Pasch, A.; Bakker, S.J.L.; De Borst, M.H.; Hoenderop, J.G.J.; De Baaij, J.H. Calciprotein particle inhibition explains magnesium-mediated protection against vascular calcification. Nephrol. Dial. Transplant. 2019. [Google Scholar] [CrossRef]
  20. Nakazato, J.; Hoshide, S.; Wake, M.; Miura, Y.; Kuro-O, M.; Kario, K. Association of calciprotein particles measured by a new method with coronary artery plaque in patients with coronary artery disease: A cross-sectional study. J. Cardiol. 2019, 74, 428–435. [Google Scholar] [CrossRef]
  21. Nakamura, K.; Nagata, Y.; Hiroyoshi, T.; Isoyama, N.; Fujikawa, K.; Miura, Y.; Matsuyama, H.; Kuro-O, M. The effect of lanthanum carbonate on calciprotein particles in hemodialysis patients. Clin. Exp. Nephrol. 2019, 24, 323–329. [Google Scholar] [CrossRef] [Green Version]
  22. Nemoto, Y.; Kumagai, T.; Ishizawa, K.; Miura, Y.; Shiraishi, T.; Morimoto, C.; Sakai, K.; Omizo, H.; Yamazaki, O.; Tamura, Y.; et al. Phosphate binding by sucroferric oxyhydroxide ameliorates renal injury in the remnant kidney model. Sci. Rep. 2019, 9, 1732. [Google Scholar] [CrossRef] [Green Version]
  23. Chen, W.; Anokhina, V.; Dieudonne, G.; Abramowitz, M.K.; Kashyap, R.; Yan, C.; Wu, T.T.; Bentley, K.L.D.M.; Miller, B.L.; Bushinsky, D. Patients with advanced chronic kidney disease and vascular calcification have a large hydrodynamic radius of secondary calciprotein particles. Nephrol. Dial. Transplant. 2019, 34, 992–1000. [Google Scholar] [CrossRef]
  24. Cai, M.M.X.; Smith, E.R.; Kent, A.; Huang, L.; Hewitson, T.; McMahon, L.P.; Holt, S.G. Calciprotein Particle Formation in Peritoneal Dialysis Effluent is Dependent on Dialysate Calcium Concentration. Perit. Dial. Int. 2018, 38, 286–292. [Google Scholar] [CrossRef]
  25. Ruderman, I.; Smith, E.R.; Toussaint, N.D.; Hewitson, T.; Holt, S.G. Longitudinal changes in bone and mineral metabolism after cessation of cinacalcet in dialysis patients with secondary hyperparathyroidism. BMC Nephrol. 2018, 19, 113. [Google Scholar] [CrossRef] [Green Version]
  26. Aghagolzadeh, P.; Radpour, R.; Bachtler, M.; Van Goor, H.; Smith, E.R.; Lister, A.; Odermatt, A.; Feelisch, M.; Pasch, A. Hydrogen sulfide attenuates calcification of vascular smooth muscle cells via KEAP1/NRF2/NQO1 activation. Atherosclerosis 2017, 265, 78–86. [Google Scholar] [CrossRef] [Green Version]
  27. Cai, M.M.X.; Smith, E.R.; Tan, S.-J.; Hewitson, T.; Holt, S.G. The Role of Secondary Calciprotein Particles in the Mineralisation Paradox of Chronic Kidney Disease. Calcif. Tissue Int. 2017, 101, 570–580. [Google Scholar] [CrossRef]
  28. Yamada, H.; Kuro-O, M.; Ishikawa, S.-E.; Funazaki, S.; Kusaka, I.; Kakei, M.; Hara, K. Daily variability in serum levels of calciprotein particles and their association with mineral metabolism parameters: A cross-sectional pilot study. Nephrology 2018, 23, 226–230. [Google Scholar] [CrossRef]
  29. Aghagolzadeh, P.; Bachtler, M.; Bijarnia, R.; Jackson, C.B.; Smith, E.R.; Odermatt, A.; Radpour, R.; Pasch, A. Calcification of vascular smooth muscle cells is induced by secondary calciprotein particles and enhanced by tumor necrosis factor-α. Atherosclerosis 2016, 251, 404–414. [Google Scholar] [CrossRef] [Green Version]
  30. Cai, M.M.X.; Wigg, B.; Smith, E.R.; Hewitson, T.; McMahon, L.P.; Holt, S.G. Relative abundance of fetuin- A in peritoneal dialysis effluent and its association with in situ formation of calciprotein particles: An observational pilot study. Nephrology 2014, 20, 6–10. [Google Scholar] [CrossRef]
  31. Smith, E.R.; Hanssen, E.; McMahon, L.P.; Holt, S.G. Fetuin-A-Containing Calciprotein Particles Reduce Mineral Stress in the Macrophage. PLoS ONE 2013, 8, e60904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Smith, E.R.; Cai, M.M.; McMahon, L.P.; Pedagogos, E.; Toussaint, N.D.; Brumby, C.; Holt, S.G. Serum fetuin-A concentration and fetuin-A-containing calciprotein particles in patients with chronic inflammatory disease and renal failure. Nephrology 2013, 18, 215–221. [Google Scholar] [CrossRef] [PubMed]
  33. Smith, E.R.; Ford, M.L.; Tomlinson, L.; Rajkumar, C.; McMahon, L.P.; Holt, S.G. Phosphorylated fetuin-A-containing calciprotein particles are associated with aortic stiffness and a procalcific milieu in patients with pre-dialysis CKD. Nephrol. Dial. Transplant. 2012, 27, 1957–1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ter Meulen, K.J.; Dekker, M.J.E.; Pasch, A.; Broers, N.J.H.; Van Der Sande, F.M.; Kooman, J.P.; Konings, C.J.A.M.; Gsponer, I.M.; Bachtler, M.D.N.; Gauly, A.; et al. Citric-acid dialysate improves the calcification propensity of hemodialysis patients: A multicenter prospective randomized cross-over trial. PLoS ONE 2019, 14, e0225824. [Google Scholar] [CrossRef] [Green Version]
  35. Van Dijk, P.R.; Hop, H.; Waanders, F.; Mulder, U.J.; Pasch, A.; Hillebrands, J.-L.; Van Goor, H.; Bilo, H.J. Serum calcification propensity in type 1 diabetes associates with mineral stress. Diabetes Res. Clin. Pract. 2019, 158, 107917. [Google Scholar] [CrossRef]
  36. Bundy, J.D.; Cai, X.; Mehta, R.C.; Scialla, J.J.; De Boer, I.H.; Hsu, C.-Y.; Go, A.S.; Dobre, M.; Chen, J.; Rao, P.S.; et al. Serum Calcification Propensity and Clinical Events in CKD. Clin. J. Am. Soc. Nephrol. 2019, 14, 1562–1571. [Google Scholar] [CrossRef]
  37. Ponte, B.; Pruijm, M.; Pasch, A.; Dufey-Teso, A.; Martin, P.-Y.; De Seigneux, S. Dialysis initiation improves calcification propensity. Nephrol. Dial. Transplant. 2019, 35, 495–502. [Google Scholar] [CrossRef]
  38. Bundy, J.D.; Cai, X.; Scialla, J.J.; Dobre, M.A.; Chen, J.; Hsu, C.-Y.; Leonard, M.B.; Go, A.S.; Rao, P.S.; Lash, J.P.; et al. Serum Calcification Propensity and Coronary Artery Calcification Among Patients With CKD: The CRIC (Chronic Renal Insufficiency Cohort) Study. Am. J. Kidney Dis. 2019, 73, 806–814. [Google Scholar] [CrossRef]
  39. Quiñones, H.; Hamdi, T.; Sakhaee, K.; Pasch, A.; Moe, O.W.; Pak, C.Y.C. Control of metabolic predisposition to cardiovascular complications of chronic kidney disease by effervescent calcium magnesium citrate: A feasibility study. J. Nephrol. 2018, 32, 93–100. [Google Scholar] [CrossRef] [Green Version]
  40. Aigner, C.; Cejka, D.; Sliber, C.; Fraunschiel, M.; Sunder-Plassmann, G.; Gaggl, M. Oral Sodium Bicarbonate Supplementation Does Not Affect Serum Calcification Propensity in Patients with Chronic Kidney Disease and Chronic Metabolic Acidosis. Kidney Blood Press. Res. 2019, 44, 188–199. [Google Scholar] [CrossRef]
  41. Bullen, A.L.; Anderson, C.A.M.; Hooker, E.R.; Kado, D.M.; Orwoll, E.; Pasch, A.; Ix, J.H. Correlates of T50 and relationships with bone mineral density in community-living older men: The osteoporotic fractures in men (MrOS) study. Osteoporos. Int. 2019, 30, 1529–1531. [Google Scholar] [CrossRef] [PubMed]
  42. Bostom, A.; Pasch, A.; Madsen, T.; Roberts, M.B.; Franceschini, N.; Steubl, D.; Garimella, P.S.; Ix, J.H.; Tuttle, K.R.; Ivanova, A.; et al. Serum Calcification Propensity and Fetuin-A: Biomarkers of Cardiovascular Disease in Kidney Transplant Recipients. Am. J. Nephrol. 2018, 48, 21–31. [Google Scholar] [CrossRef] [PubMed]
  43. Andrews, E.S.; Perrenoud, L.; Nowak, K.L.; You, Z.; Pasch, A.; Chonchol, M.; Kendrick, J.; Jalal, D. Examining the effects of uric acid-lowering on markers vascular of calcification and CKD-MBD. A post-hoc analysis of a randomized clinical trial. PLoS ONE 2018, 13, e0205831. [Google Scholar] [CrossRef] [PubMed]
  44. Ussif, A.M.; Pihlstrøm, H.; Pasch, A.; Holdaas, H.; Hartmann, A.; Smerud, K.; Åsberg, A. Paricalcitol supplementation during the first year after kidney transplantation does not affect calcification propensity score. BMC Nephrol. 2018, 19, 212. [Google Scholar] [CrossRef]
  45. Bressendorff, I.; Hansen, D.; Schou, M.; Pasch, A.; Brandi, L. The Effect of Increasing Dialysate Magnesium on Serum Calcification Propensity in Subjects with End Stage Kidney Disease. Clin. J. Am. Soc. Nephrol. 2018, 13, 1373–1380. [Google Scholar] [CrossRef] [Green Version]
  46. Lorenz, G.; Mayer, C.C.; Bachmann, Q.; Stryeck, S.; Braunisch, M.C.; Haller, B.; Carbajo-Lozoya, J.; Schmidt, A.; Witthauer, S.; Abuzahu, J.; et al. Acetate-free, citrate-acidified bicarbonate dialysis improves serum calcification propensity—a preliminary study. Nephrol. Dial. Transplant. 2018, 33, 2043–2051. [Google Scholar] [CrossRef]
  47. Dahdal, S.; Devetzis, V.; Chalikias, G.; Tziakas, D.; Chizzolini, C.; Ribi, C.; Trendelenburg, M.; Eisenberger, U.; Hauser, T.; Pasch, A.; et al. Serum calcification propensity is independently associated with disease activity in systemic lupus erythematosus. PLoS ONE 2018, 13, e0188695. [Google Scholar] [CrossRef] [Green Version]
  48. Pruijm, M.; Lu, Y.; Megdiche, F.; Piskunowicz, M.; Milani, B.; Stuber, M.; Bachtler, M.; Vogt, B.; Burnier, M.; Pasch, A. Serum calcification propensity is associated with renal tissue oxygenation and resistive index in patients with arterial hypertension or chronic kidney disease. J. Hypertens. 2017, 35, 2044–2052. [Google Scholar] [CrossRef]
  49. Pasch, A.; Block, G.A.; Bachtler, M.; Smith, E.R.; Jahnen-Dechent, W.; Arampatzis, S.; Chertow, G.M.; Parfrey, P.; Ma, X.; Floege, J. Blood Calcification Propensity, Cardiovascular Events, and Survival in Patients Receiving Hemodialysis in the EVOLVE Trial. Clin. J. Am. Soc. Nephrol. 2016, 12, 315–322. [Google Scholar] [CrossRef] [Green Version]
  50. Bielesz, B.; Reiter, T.; Marculescu, R.; Gleiss, A.; Bojic, M.; Kieweg, H.; Cejka, D. Calcification Propensity of Serum is Independent of Excretory Renal Function. Sci. Rep. 2017, 7, 17941. [Google Scholar] [CrossRef] [Green Version]
  51. Bressendorff, I.; Hansen, D.; Schou, M.; Silver, B.; Pasch, A.; Bouchelouche, P.; Pedersen, L.; Rasmussen, L.M.; Brandi, L. Oral Magnesium Supplementation in Chronic Kidney Disease Stages 3 and 4: Efficacy, Safety, and Effect on Serum Calcification Propensity-A Prospective Randomized Double-Blinded Placebo-Controlled Clinical Trial. Kidney Int. Rep. 2016, 2, 380–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Lorenz, G.; Steubl, D.; Kemmner, S.; Pasch, A.; Koch-Sembdner, W.; Pham, D.; Haller, B.; Bachmann, Q.; Mayer, C.C.; Wassertheurer, S.; et al. Worsening calcification propensity precedes all-cause and cardiovascular mortality in haemodialyzed patients. Sci. Rep. 2017, 7, 13368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Smerud, K.; Åsberg, A.; Kile, H.; Pasch, A.; Dahle, D.O.; Bollerslev, J.; Godang, K.; Hartmann, A. A rapid and sustained improvement of calcification propensity score (serum T50) after successful kidney transplantation: Reanalysis of a randomized controlled trial of ibandronate. Clin. Transplant. 2017, 31, e13131. [Google Scholar] [CrossRef] [PubMed]
  54. Dekker, M.; Pasch, A.; Van Der Sande, F.; Konings, C.; Bachtler, M.; Dionisi, M.; Meier, M.; Kooman, J.; Canaud, B. High-Flux Hemodialysis and High-Volume Hemodiafiltration Improve Serum Calcification Propensity. PLoS ONE 2016, 11, e0151508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bristow, S.; Gamble, G.D.; Pasch, A.; O’Neill, W.C.; Stewart, A.; Horne, A.; Reid, I.R. Acute and 3-month effects of calcium carbonate on the calcification propensity of serum and regulators of vascular calcification: Secondary analysis of a randomized controlled trial. Osteoporos. Int. 2015, 27, 1209–1216. [Google Scholar] [CrossRef] [PubMed]
  56. Dahle, D.O.; Åsberg, A.; Hartmann, A.; Holdaas, H.; Bachtler, M.; Jenssen, T.G.; Dionisi, M.; Pasch, A. Serum Calcification Propensity Is a Strong and Independent Determinant of Cardiac and All-Cause Mortality in Kidney Transplant Recipients. Arab. Archaeol. Epigr. 2015, 16, 204–212. [Google Scholar] [CrossRef] [Green Version]
  57. Keyzer, C.A.; De Borst, M.H.; Berg, E.V.D.; Jahnen-Dechent, W.; Arampatzis, S.; Farese, S.; Bergmann, I.P.; Floege, J.; Navis, G.; Bakker, S.J.; et al. Calcification Propensity and Survival among Renal Transplant Recipients. J. Am. Soc. Nephrol. 2015, 27, 239–248. [Google Scholar] [CrossRef] [Green Version]
  58. Berchtold, L.; Ponte, B.; Moll, S.; Hadaya, K.; Seyde, O.; Bachtler, M.; Vallée, J.-P.; Martin, P.-Y.; Pasch, A.; De Seigneux, S. Phosphocalcic Markers and Calcification Propensity for Assessment of Interstitial Fibrosis and Vascular Lesions in Kidney Allograft Recipients. PLoS ONE 2016, 11, e0167929. [Google Scholar] [CrossRef]
  59. De Seigneux, S.; Ponte, B.; Berchtold, L.; Hadaya, K.; Martin, P.-Y.; Pasch, A. Living kidney donation does not adversely affect serum calcification propensity and markers of vascular stiffness. Transpl. Int. 2015, 28, 1074–1080. [Google Scholar] [CrossRef]
  60. Smith, E.R.; Ford, M.L.; Tomlinson, L.; Bodenham, E.; McMahon, L.P.; Farese, S.; Rajkumar, C.; Holt, S.G.; Pasch, A. Serum Calcification Propensity Predicts All-Cause Mortality in Predialysis CKD. J. Am. Soc. Nephrol. 2013, 25, 339–348. [Google Scholar] [CrossRef]
  61. Zarb, Y.; Weber-Stadlbauer, U.; Kirschenbaum, D.; Kindler, D.R.; Richetto, J.; Keller, D.; Rademakers, R.; Dickson, D.W.; Pasch, A.; Byzova, T.V.; et al. Ossified blood vessels in primary familial brain calcification elicit a neurotoxic astrocyte response. Brain 2019, 142, 885–902. [Google Scholar] [CrossRef] [PubMed]
  62. Kendrick, J.; Shah, P.; Andrews, E.; You, Z.; Nowak, K.L.; Pasch, A.; Chonchol, M. Effect of Treatment of Metabolic Acidosis on Vascular Endothelial Function in Patients with CKD. Clin. J. Am. Soc. Nephrol. 2018, 13, 1463–1470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Voelkl, J.; Tuffaha, R.; Luong, T.T.; Zickler, D.; Masyout, J.; Feger, M.; Verheyen, N.; Blaschke, F.; Kuro-O, M.; Tomaschitz, A.; et al. Zinc Inhibits Phosphate-Induced Vascular Calcification through TNFAIP3-Mediated Suppression of NF-κB. J. Am. Soc. Nephrol. 2018, 29, 1636–1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jahnen-Dechent, W.; Trindl, A.; Godovac-Zimmermann, J.; Müller-Esterl, W. Posttranslational Processing of Human alpha2-HS Glycoprotein (Human Fetuin). Evidence for the Production of a Phosphorylated Single-Chain Form by Hepatoma Cells. JBIC J. Boil. Inorg. Chem. 1994, 226, 59–69. [Google Scholar] [CrossRef] [PubMed]
  65. Mathews, S.T.; Chellam, N.; Srinivas, P.R.; Cintron, V.J.; Leon, M.; Goustin, A.S.; Grunberger, G. Alpha2-HSG, a specific inhibitor of insulin receptor autophosphorylation, interacts with the insulin receptor. Mol. Cell. Endocrinol. 2000, 164, 87–98. [Google Scholar] [CrossRef]
  66. Auberger, P.; Falquerho, L.; Contreres, J.O.; Pagès, G.; Le Cam, G.; Rossi, B.; Le Cam, A. Characterization of a natural inhibitor of the insulin receptor tyrosine kinase: cDNA cloning, purification, and anti-mitogenic activity. Cell 1989, 58, 631–640. [Google Scholar] [CrossRef]
  67. Suzuki, M.; Shimokawa, H.; Takagi, Y.; Sasaki, S. Calcium-binding properties of fetuin in fetal bovine serum. J. Exp. Zoöl. 1994, 270, 501–507. [Google Scholar] [CrossRef]
  68. Herrmann, M.; Schäfer, C.; Heiss, A.; Gräber, S.; Kinkeldey, A.; Büscher, A.; Schmitt, M.M.; Bornemann, J.; Nimmerjahn, F.; Herrmann, M.; et al. Clearance of Fetuin-A–Containing Calciprotein Particles Is Mediated by Scavenger Receptor-A. Circ. Res. 2012, 111, 575–584. [Google Scholar] [CrossRef]
  69. Hamano, T.; Matsui, I.; Mikami, S.; Tomida, K.; Fujii, N.; Imai, E.; Rakugi, H.; Isaka, Y. Fetuin-mineral complex reflects extraosseous calcification stress in CKD. J. Am. Soc. Nephrol. 2010, 21, 1998–2007. [Google Scholar] [CrossRef]
  70. Smith, E.R.; Hewitson, T.; Cai, M.M.X.; Aghagolzadeh, P.; Bachtler, M.; Pasch, A.; Holt, S.G. A novel fluorescent probe-based flow cytometric assay for mineral-containing nanoparticles in serum. Sci. Rep. 2017, 7, 5686. [Google Scholar] [CrossRef] [Green Version]
  71. Ismail, A.H.; Schäfer, C.; Heiss, A.; Walter, M.; Jahnen-Dechent, W.; Leonhardt, S. An electrochemical impedance spectroscopy (EIS) assay measuring the calcification inhibition capacity in biological fluids. Biosens. Bioelectron. 2011, 26, 4702–4707. [Google Scholar] [CrossRef] [PubMed]
  72. Ter Braake, A.D.; Tinnemans, P.T.; Shanahan, C.M.; Hoenderop, J.G.J.; De Baaij, J.H. Magnesium prevents vascular calcification in vitro by inhibition of hydroxyapatite crystal formation. Sci. Rep. 2018, 8, 2069. [Google Scholar] [CrossRef] [PubMed]
  73. Huang, M.; Zheng, L.; Xu, H.; Tang, D.; Lin, L.; Zhang, J.; Li, C.; Wang, W.; Yuan, Q.; Tao, L.; et al. Oxidative stress contributes to vascular calcification in patients with chronic kidney disease. J. Mol. Cell. Cardiol. 2020, 138, 256–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Rubin, M.R.; Silverberg, S.J. Vascular calcification and osteoporosis--the nature of the nexus. J. Clin. Endocrinol. Metab. 2004, 89, 4243–4245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Keeley, F.; Sitarz, E. Identification and quantitation of α2-HS-glycoprotein in the mineralized matrix of calcified plaques of atherosclerotic human aorta. Atherosclerosis 1985, 55, 63–69. [Google Scholar] [CrossRef]
  76. Binkert, C.; Demetriou, M.; Sukhu, B.; Szweras, M.; Tenenbaum, H.C.; Dennis, J. Regulation of osteogenesis by fetuin. J. Boil. Chem. 1999, 274, 28514–28520. [Google Scholar] [CrossRef] [Green Version]
  77. Ketteler, M.; Bongartz, P.; Westenfeld, R.; Wildberger, J.E.; Mahnken, A.H.; Böhm, R.; Metzger, T.; Wanner, C.; Jahnen-Dechent, W.; Floege, J. Association of low fetuin-A (AHSG) concentrations in serum with cardiovascular mortality in patients on dialysis: A cross-sectional study. Lancet 2003, 361, 827–833. [Google Scholar] [CrossRef]
  78. Fontaine, M.A.; Albert, A.; Dubois, B.; Saint-Remy, A.; Rorive, G. Fracture and bone mineral density in hemodialysis patients. Clin. Nephrol. 2000, 54, 218–226. [Google Scholar]
  79. Kirkpantur, A.; Altun, B.; Hazirolan, T.; Akata, D.; Arici, M.; Kirazli, S.; Turgan, C. Association Among Serum Fetuin-A Level, Coronary Artery Calcification, and Bone Mineral Densitometry in Maintenance Hemodialysis Patients. Artif. Organs 2009, 33, 844–854. [Google Scholar] [CrossRef]
  80. Pasch, A.; Jahnen-Dechent, W.; Smith, E.R. Phosphate, Calcification in Blood, and Mineral Stress: The Physiologic Blood Mineral Buffering System and Its Association with Cardiovascular Risk. Int. J. Nephrol. 2018, 2018, 1–5. [Google Scholar] [CrossRef]
Figure 1. Flow diagram for the identification, selection, screening, and inclusion process. Abbreviations: CPP, calciprotein particles.
Figure 1. Flow diagram for the identification, selection, screening, and inclusion process. Abbreviations: CPP, calciprotein particles.
Jcm 09 01287 g001
Figure 2. Fetuin-A transformation into CPP II. Abbreviations: Pi, phosphate; CPP I, primary calciprotein particle; CPP II, secondary calciprotein particle.
Figure 2. Fetuin-A transformation into CPP II. Abbreviations: Pi, phosphate; CPP I, primary calciprotein particle; CPP II, secondary calciprotein particle.
Jcm 09 01287 g002
Figure 3. Effects of various factors upon T50 (half of the time needed for the spontaneous transition from CPP I to CPP II) and plasma calcification propensity. Abbreviations: Pi, phosphate; Ca2+, calcium; Mg2+, magnesium.
Figure 3. Effects of various factors upon T50 (half of the time needed for the spontaneous transition from CPP I to CPP II) and plasma calcification propensity. Abbreviations: Pi, phosphate; Ca2+, calcium; Mg2+, magnesium.
Jcm 09 01287 g003
Figure 4. Summary of factors that increase T50 in CKD patients. Abbreviations: Mg, magnesium; VC, vascular calcification.
Figure 4. Summary of factors that increase T50 in CKD patients. Abbreviations: Mg, magnesium; VC, vascular calcification.
Jcm 09 01287 g004
Table 1. Summary of 11 human studies on calciprotein particle (CPP).
Table 1. Summary of 11 human studies on calciprotein particle (CPP).
Author, YearStudy Design, DurationNumber of Subjects, DiseaseCPP Type StudiedFindings
Nakazato et al. 2019 [20]cross-sectional, N/A71 ACStCPPHigh CPP levels associated with atherosclerosis.
Chen et al. 2019 [23]cross-sectional, N/A45 CKD stage IV–VCPP IILarger CPP II diameter in patients with VC.
Viegas et al. 2018 [3]cross-sectional, N/A16 CKD stage II-IV, 20 CKD stage VtCPPCPP from CKD stage V patients contained less fetuin-A and GRP and had CPP II like characteristics.
Yamada et al. 2018 [28]cross-sectional, N/A10 diabetes mellitus type 2tCPPCPP elevated 2 h post-meal, CPP inversely correlated with eGFR.
Cai et al. 2015 [30]cross-sectional, N/A20 peritoneal dialysistCPPCPP present, fetuin-A abundant in peritoneal dialysis effluent.
Smith et al. 2013 [32]cross-sectional, N/A11 CKD stage III–IV, 42 HD, 18 peritoneal dialysis, 13 chronic inflammatory diseasetCPPCPP increased in CKD III-IV, HD, peritoneal dialysis and chronic inflammatory disease patients; CPP was highest in HD patients with calcific uremic arteriolopathy.
Smith et al. 2012 [33]cross-sectional, N/A200 CKD stage III–IVtCPPHigher CPP levels associated with increased aortic stiffness.
Cai et al. 2018 [24]prospective cohort, 7 weeks12 peritoneal dialysistCPPDialysate with higher Ca2+ concentration had higher CPP content.
Ruderman et al. 2018 [25]prospective cohort, 12 months62 HDCPP IIncrease of serum CPP I after cessation of cinacalcet treatment.
Bressendorff et al. 2019 [17]Interventional, 28 days57 HDCPP I, CPP IIHigher Mg2+ concentration dialysis solution reduced both CPP I and CPP II levels, compared to standard dialysis solution.
Nakamura et al. 2019 [21]Interventional, 16 weeks24 HDtCPPLower CPP in lanthanum carbonate treated patients vs. calcium carbonate.
Abbreviations: HD, haemodialysis; ACS, acute coronary syndrome; CPP, calciprotein particle; CPP I, primary calciprotein particle; CPP II, secondary calciprotein particle; tCPP, total calciprotein particles; CKD, chronic kidney disease; VC, vascular calcification; GRP, Gla-rich protein; eGFR, estimated glomerular filtration rate; N/A, not applicable.
Table 2. Summary of 1 animal and 6 in vitro studies on CPP.
Table 2. Summary of 1 animal and 6 in vitro studies on CPP.
Author, YearStudy DesignAnimals/CellsCPP Type StudiedFindings
Nemoto et al. 2019 [22]animalrats with 5/6 nephrectomytCPPLower CPP in rats treated with sucroferric oxyhydroxide.
Shishkova et al. 2019 [18]in vitroVSMCsCPP I, CPP IIBoth CPP I and CPP II induced VSMC intimal hyperplasia, more pronounced in case of CPP II.
Ter Braake et al. 2019 [19]in vitroVSMCsCPP IICPP II induced VSMC calcification.
Aghagolzadeh et al. 2017 [26]in vitroVSMCstCPPH2S inhibits CPP induced VSMC calcification.
Cai et al. 2017 [27]in vitroVSMCsCPP IIPi or CPP II alone did not initiate VSMC mineralization, but CPP II with Pi did.
Aghagolzadeh et al. 2016 [29]in vitroVSMCsCPP I, CPP IICPP II induced calcification in VSMCs, CPP I did not.
Smith et al. 2013 [31]in vitroVSMCstCPPCPP induce secretion of TNF-α and IL-1β in macrophages, but less significantly than that induced by hydroxyapatite crystals.
Abbreviations: VSMCs, vascular smooth muscle cells; CPP, calciprotein particle; CPP I, primary calciprotein particle; CPP II, secondary calciprotein particle; tCPP, total calciprotein particles; H2S, hydrogen sulphide; Pi, phosphate; TNF-α, tumour necrosis factor α; IL-1β, interleukin 1β.
Table 3. Summary of 18 observational studies on T50 and health outcomes.
Table 3. Summary of 18 observational studies on T50 and health outcomes.
Author, YearStudy DesignFollow-Up TimeNumber of Subjects, DiseaseFindings
Bullen et al. 2019 [41]cross-sectionalN/A149 men with osteoporosisT50 was not associated with bone mineral density.
Dahdal et al. 2018 [47]cross-sectionalN/A168, SLET50 was negatively associated with disease activity.
Pruijm et al. 2017 [48]cross-sectionalN/A58, CKD; 48, hypertensionLower T50 was associated with reduced tissue oxygenation and perfusion.
Bielesz et al. 2017 [50]cross-sectionalN/A118, CKD stage I–VT50 associated with Pi, Mg2+ and fetuin-A but not with eGFR.
Dekker et al. 2016 [54]cross-sectionalN/A64, HDT50 increased post-haemodialysis and post-haemodiafiltration.
Voelkl et al. 2018 [63]cross-sectionalN/A16, CKD; 20, HDT50 was lower in CKD patients compared to controls.
van Dijk et al. 2019 [35]prospective cohort15 years216, type 1 diabetesT50 not associated with mortality.
Bundy et al. 2019 [36]prospective cohortAt TOD or 11.2 years3404, CKD stage II–IVLower T50 associated with cardiovascular events and all-cause mortality.
Ponte et al. 2019 [37]prospective cohort3 months46, HD; 12, peritoneal dialysisHigher T 50 after dialysis initiation.
Bundy et al. 2019 [38]prospective cohort3.2 ± 0.6 years780, CKD stage II–IVLower T50 was associated with greater CAC severity and progression, however, T50 was not associated with CAC incidence.
Bostom et al. 2018 [42]prospective cohortmedian of 2.18 years685, CVDLower T50 and fetuin-A levels were associated with greater risk for CVD outcomes.
Pasch et al. 2017 [49]prospective cohortAt TOD or first non-fatal CVE2785, HDLower T50 associated with all-cause mortality, myocardial infarction, and peripheral vascular events.
Lorenz et al. 2017 [52]prospective cohort24 months188, HDT50 rate of decline significantly predicted all-cause and cardiovascular mortality.
Dahle et al. 2016 [56]prospective cohortmedian of 5.1 years1435, kidney transplantLower T50 associated with all-cause and cardiac mortality.
Keyzer et al. 2016 [57]prospective cohortmedian of 3.1 years699, kidney transplantLower T50 associated with increased graft failure, all-cause, and cardiac mortality.
de Seigneux et al. 2015 [59]prospective cohort1 year21, kidney donorsT50 was independent of eGFR.
Smith et al. 2014 [60]prospective cohortmedian of 5.3 years184, CKD stage III–IVLower T50 associated with higher all-cause mortality.
Berchtold et al. 2016 [58]retrospective cohortbetween 2 and 43 years129, kidney transplantT50 associated with interstitial fibrosis and vascular lesions.
Abbreviations: SLE, systemic lupus erythematosus; HD, haemodialysis; CKD, chronic kidney disease; CAC, coronary artery calcification; CVD, cardiovascular disease; Mg2+, magnesium; TOD, time of death; CVE, cardiovascular event; Pi, phosphate; eGFR, estimated glomerular filtration rate; N/A, not applicable.
Table 4. Summary of 11 human interventional studies on T50 with health outcomes.
Table 4. Summary of 11 human interventional studies on T50 with health outcomes.
Author, YearStudy DurationNumber of Subjects, DiseaseFindings
Smerud et al. 2017 [53]1 year123, kidney transplantT50 increased with no further change after 1 year, ibandronate had no effect on T50.
Andrews et al. 2018 [43]12 weeks80, CKD with hyperuricemiaAllopurinol lowered uric acid levels but had no effect on T50.
Lorenz et al. 2018 [46]3 months78, HDAcetate-free, citrate-acidified, standard bicarbonate dialysis solution increased T50 compared to acetate dialysis solution.
Ussif et al. 2018 [44]1 year76, kidney transplantParicalcitol supplementation had no effect on T50.
Bressendorff et al. 2018 [45]28 days57, HDHigher dialysis solution Mg2+ concentration increased T50.
Bristow et al. 2016 [55]3 months41, post-menopausal womenInsignificant decrease of T50 in the group treated with oral calcium carbonate supplement.
Bressendorff et al. 2017 [51]8 weeks36, CKD III–IVOral Mg2+ supplementation increased T50.
Aigner et al. 2019 [40]4 weeks35, CKDOral bicarbonate supplementation showed no effect on T50 in acidotic CKD patients.
Kendrick et al. 2018 [62]14 weeks18, CKDOral sodium bicarbonate supplementation showed no effect on T50 in CKD patients with low serum bicarbonate levels.
Ter Meulen et al. 2019 [34]2 weeks18, HDCitric acid-buffered dialysis solution increased T50 compared to acetate-buffered solution.
Quiñones et al. 2019 [39]2 weeks9, CKD stage III, 9, CKD stage VEffervescent, oral, calcium-magnesium citrate increased T50.
Abbreviations: HD, haemodialysis; CKD, chronic kidney disease; Mg2+, magnesium.

Share and Cite

MDPI and ACS Style

Silaghi, C.N.; Ilyés, T.; Van Ballegooijen, A.J.; Crăciun, A.M. Calciprotein Particles and Serum Calcification Propensity: Hallmarks of Vascular Calcifications in Patients with Chronic Kidney Disease. J. Clin. Med. 2020, 9, 1287. https://doi.org/10.3390/jcm9051287

AMA Style

Silaghi CN, Ilyés T, Van Ballegooijen AJ, Crăciun AM. Calciprotein Particles and Serum Calcification Propensity: Hallmarks of Vascular Calcifications in Patients with Chronic Kidney Disease. Journal of Clinical Medicine. 2020; 9(5):1287. https://doi.org/10.3390/jcm9051287

Chicago/Turabian Style

Silaghi, Ciprian N., Tamás Ilyés, Adriana J. Van Ballegooijen, and Alexandra M. Crăciun. 2020. "Calciprotein Particles and Serum Calcification Propensity: Hallmarks of Vascular Calcifications in Patients with Chronic Kidney Disease" Journal of Clinical Medicine 9, no. 5: 1287. https://doi.org/10.3390/jcm9051287

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop